Syncytial Mutations Do Not Impair the Specificity of Entry and Spread of a Glycoprotein D Receptor-Retargeted Herpes Simplex Virus

J Virol. 2016 Nov 28;90(24):11096-11105. doi: 10.1128/JVI.01456-16. Print 2016 Dec 15.

Abstract

Membrane fusion, which is the key process for both initial cell entry and subsequent lateral spread of herpes simplex virus (HSV), requires the four envelope glycoproteins gB, gD, gH, and gL. Syncytial mutations, predominantly mapped to the gB and gK genes, confer hyperfusogenicity on HSV and cause multinucleated giant cells, termed syncytia. Here we asked whether interaction of gD with a cognate entry receptor remains indispensable for initiating membrane fusion of syncytial strains. To address this question, we took advantage of mutant viruses whose viral entry into cells relies on the uniquely specific interaction of an engineered gD with epidermal growth factor receptor (EGFR). We introduced selected syncytial mutations into gB and/or gK of the EGFR-retargeted HSV and found that these mutations, especially when combined, enabled formation of extensive syncytia by human cancer cell lines that express the target receptor; these syncytia were substantially larger than the plaques formed by the parental retargeted HSV strain. We assessed the EGFR dependence of entry and spread separately by using direct entry and infectious center assays, respectively, and we found that the syncytial mutations did not override the receptor specificity of the retargeted viruses at either stage. We discuss the implications of these results for the development of more effective targeted oncolytic HSV vectors.

Importance: Herpes simplex virus (HSV) is investigated not only as a human pathogen but also as a promising agent for oncolytic virotherapy. We previously showed that both the initial entry and subsequent lateral spread of HSV can be retargeted to cells expressing tumor-associated antigens by single-chain antibodies fused to a receptor-binding-deficient envelope glycoprotein D (gD). Here we introduced syncytial mutations into the gB and/or gK gene of gD-retargeted HSVs to determine whether viral tropism remained dependent on the interaction of gD with the target receptor. Entry and spread profiles of the recombinant viruses indicated that gD retargeting does not abolish the hyperfusogenic activity of syncytial mutations and that these mutations do not eliminate the dependence of HSV entry and spread on a specific gD-receptor interaction. These observations suggest that syncytial mutations may be valuable for increasing the tumor-specific spreading of retargeted oncolytic HSV vectors.

MeSH terms

  • Animals
  • CHO Cells
  • Cell Line, Tumor
  • Cell Survival
  • Chlorocebus aethiops
  • Cricetulus
  • ErbB Receptors / genetics
  • ErbB Receptors / metabolism*
  • Gene Expression
  • Giant Cells / metabolism
  • Giant Cells / ultrastructure
  • Giant Cells / virology
  • Herpesvirus 1, Human / genetics*
  • Herpesvirus 1, Human / metabolism
  • Host-Pathogen Interactions
  • Humans
  • Membrane Fusion
  • Mutagenesis, Site-Directed
  • Mutation*
  • Oncolytic Virotherapy
  • Receptors, Virus / genetics
  • Receptors, Virus / metabolism*
  • Vero Cells
  • Viral Envelope Proteins / genetics*
  • Viral Envelope Proteins / metabolism
  • Virus Internalization

Substances

  • Receptors, Virus
  • Viral Envelope Proteins
  • glycoprotein D, Human herpesvirus 1
  • EGFR protein, human
  • ErbB Receptors

Grants and funding

This work, including the efforts of Hiroaki Uchida, was funded by Japan Society for the Promotion of Science (JSPS) (25290059 and 15K15144), Takeda Science Foundation, Mochida Memorial Foundation for Medical and Pharmaceutical Research, Daiwa Securities Health Foundation, and Sumitomo Foundation. The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.